Modulating of ocular inflammation with macrophage migration inhibitory factor is associated with notch signalling in experimental autoimmune uveitis

Clin Exp Immunol. 2016 Feb;183(2):280-93. doi: 10.1111/cei.12710. Epub 2015 Nov 17.

Abstract

The aim of this study was to examine whether macrophage migration inhibitory factor (MIF) could exaggerate inflammatory response in a mouse model of experimental autoimmune uveitis (EAU) and to explore the underlying mechanism. Mutant serotype 8 adeno-associated virus (AAV8) (Y733F)-chicken β-actin (CBA)-MIF or AAV8 (Y733F)-CBA-enhanced green fluorescent protein (eGFP) vector was delivered subretinally into B10.RIII mice, respectively. Three weeks after vector delivery, EAU was induced with a subcutaneous injection of a mixture of interphotoreceptor retinoid binding protein (IRBP) peptide with CFA. The levels of proinflammatory cytokines were detected by real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA). Retinal function was evaluated with electroretinography (ERG). We found that the expression of MIF and its two receptors CD74 and CD44 was increased in the EAU mouse retina. Compared to AAV8.CBA.eGFP-injected and untreated EAU mice, the level of proinflammatory cytokines, the expression of Notch1, Notch4, delta-like ligand 4 (Dll4), Notch receptor intracellular domain (NICD) and hairy enhancer of split-1 (Hes-1) increased, but the ERG a- and b-wave amplitudes decreased in AAV8.CBA.MIF-injected EAU mice. The Notch inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT) reduced the expression of NICD, Hes-1 and proinflammatory cytokines. Further, a MIF antagonist ISO-1 attenuated intraocular inflammation, and inhibited the differentiation of T helper type 1 (Th1) and Th17 in EAU mice. We demonstrated that over-expression of MIF exaggerated ocular inflammation, which was associated with the activation of the Notch signalling. The expression of both MIF and its receptors are elevated in EAU mice. Over-expression of MIF exaggerates ocular inflammation, and this exaggerated inflammation is associated with the activation of the Notch signalling and Notch pathway. Our data suggest that the MIF-Notch axis may play an important role in the pathogenesis of EAU. Both the MIF signalling pathways may be promising targets for developing novel therapeutic interventions for uveitis.

Keywords: EAU; MIF; Notch; inflammation; retina.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Autoimmune Diseases / immunology*
  • Autoimmune Diseases / metabolism
  • Autoimmune Diseases / therapy
  • Cytokines / genetics
  • Dependovirus / genetics
  • Disease Models, Animal
  • Electroretinography
  • Enzyme-Linked Immunosorbent Assay
  • Eye Proteins / administration & dosage
  • Female
  • Genetic Vectors
  • Intramolecular Oxidoreductases / genetics
  • Intramolecular Oxidoreductases / physiology*
  • Macrophage Migration-Inhibitory Factors / genetics
  • Macrophage Migration-Inhibitory Factors / physiology*
  • Male
  • Mice
  • Mice, Inbred Strains
  • Real-Time Polymerase Chain Reaction
  • Receptors, Notch / immunology
  • Receptors, Notch / physiology*
  • Retina / immunology*
  • Retina / physiopathology
  • Retina / ultrastructure
  • Retinol-Binding Proteins / administration & dosage
  • Signal Transduction
  • Th1 Cells / immunology
  • Th17 Cells / immunology
  • Uveitis / immunology*
  • Uveitis / metabolism
  • Uveitis / physiopathology
  • Uveitis / therapy

Substances

  • Cytokines
  • Eye Proteins
  • Macrophage Migration-Inhibitory Factors
  • Receptors, Notch
  • Retinol-Binding Proteins
  • interstitial retinol-binding protein
  • Intramolecular Oxidoreductases
  • Mif protein, mouse